Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35101972

RESUMEN

Neuroinflammation is well known to be associated with neurodegenerative diseases. Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that has been implicated in neuroinflammation, but its precise cellular and molecular mechanisms remain unknown. In this study, we generated conditional knockout (CKO) mice that lack ASK1 in T cells, dendritic cells, microglia/macrophages, microglia, or astrocytes, to assess the roles of ASK1 during experimental autoimmune encephalomyelitis (EAE). We found that neuroinflammation was reduced in both the early and later stages of EAE in microglia/macrophage-specific ASK1 knockout mice, whereas only the later-stage neuroinflammation was ameliorated in astrocyte-specific ASK1 knockout mice. ASK1 deficiency in T cells and dendritic cells had no significant effects on EAE severity. Further, we found that ASK1 in microglia/macrophages induces a proinflammatory environment, which subsequently activates astrocytes to exacerbate neuroinflammation. Microglia-specific ASK1 deletion was achieved using a CX3CR1CreER system, and we found that ASK1 signaling in microglia played a major role in generating and maintaining disease. Activated astrocytes produce key inflammatory mediators, including CCL2, that further activated and recruited microglia/macrophages, in an astrocytic ASK1-dependent manner. Astrocyte-specific analysis revealed CCL2 expression was higher in the later stage compared with the early stage, suggesting a greater proinflammatory role of astrocytes in the later stage. Our findings demonstrate cell-type-specific roles of ASK1 and suggest phase-specific ASK1-dependent glial cell interactions in EAE pathophysiology. We propose glial ASK1 as a promising therapeutic target for reducing neuroinflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , MAP Quinasa Quinasa Quinasa 5/inmunología , Microglía/inmunología , Esclerosis Múltiple/inmunología , Transducción de Señal/inmunología , Animales , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/genética , Inflamación/genética , Inflamación/inmunología , MAP Quinasa Quinasa Quinasa 5/genética , Macrófagos/inmunología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Transducción de Señal/genética , Linfocitos T/inmunología
2.
J Pharmacol Sci ; 154(4): 326-333, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38485351

RESUMEN

PURPOSE: To determine whether combination of topical ripasudil and brimonidine has more effective neuroprotection on retinal ganglion cells (RGCs) following injury to axons composing the optic nerve. METHODS: Topical ripasudil, brimonidine, or mixture of both drugs were administered to adult mice after optic nerve injury (ONI). The influence of drug conditions on RGC health were evaluated by the quantifications of surviving RGCs, phosphorylated p38 mitogen-activated protein kinase (phospho-p38), and expressions of trophic factors and proinflammatory mediators in the retina. RESULTS: Topical ripasudil and brimonidine suppressed ONI-induced RGC death respectively, and mixture of both drugs further stimulated RGC survival. Topical ripasudil and brimonidine suppressed ONI-induced phospho-p38 in the whole retina. In addition, topical ripasudil suppressed expression levels of TNFα, IL-1ß and monocyte chemotactic protein-1 (MCP-1), whereas topical brimonidine increased the expression level of basic fibroblast growth factor (bFGF). CONCLUSIONS: Combination of topical ripasudil and brimonidine may enhance RGC protection by modulating multiple signaling pathways in the retina.


Asunto(s)
Isoquinolinas , Traumatismos del Nervio Óptico , Sulfonamidas , Ratones , Animales , Tartrato de Brimonidina , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/metabolismo , Neuroprotección , Combinación de Medicamentos
3.
Mol Ther ; 31(3): 810-824, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36463402

RESUMEN

Activation of neurotrophic factor signaling is a promising therapy for neurodegeneration. However, the transient nature of ligand-dependent activation limits its effectiveness. In this study, we solved this problem by inventing a system that forces membrane localization of the intracellular domain of tropomyosin receptor kinase B (iTrkB), which results in constitutive activation without ligands. Our system overcomes the small size limitation of the genome packaging in adeno-associated virus (AAV) and allows high expression of the transgene. Using AAV-mediated gene therapy in the eyes, we demonstrate that iTrkB expression enhances neuroprotection in mouse models of glaucoma and stimulates robust axon regeneration after optic nerve injury. In addition, iTrkB expression in the retina was also effective in an optic tract transection model, in which the injury site is near the superior colliculus. Regenerating axons successfully formed pathways to their brain targets, resulting in partial recovery of visual behavior. Our system may also be applicable to other trophic factor signaling pathways and lead to a significant advance in the field of gene therapy for neurotrauma and neurodegenerative disorders, including glaucoma.


Asunto(s)
Glaucoma , Células Ganglionares de la Retina , Ratones , Animales , Células Ganglionares de la Retina/metabolismo , Axones/fisiología , Regeneración Nerviosa/genética , Retina , Glaucoma/genética , Glaucoma/terapia , Glaucoma/metabolismo , Modelos Animales de Enfermedad
4.
J Biol Chem ; 295(19): 6710-6720, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32241915

RESUMEN

The DOCK-D (dedicator of cytokinesis D) family proteins are atypical guanine nucleotide exchange factors that regulate Rho GTPase activity. The family consists of Zizimin1 (DOCK9), Zizimin2 (DOCK11), and Zizimin3 (DOCK10). Functions of the DOCK-D family proteins are presently not well-explored, and the role of the DOCK-D family in neuroinflammation is unknown. In this study, we generated three mouse lines in which DOCK9 (DOCK9-/-), DOCK10 (DOCK10-/-), or DOCK11 (DOCK11-/-) had been deleted and examined the phenotypic effects of these gene deletions in MOG35-55 peptide-induced experimental autoimmune encephalomyelitis, an animal model of the neuroinflammatory disorder multiple sclerosis. We found that all the gene knockout lines were healthy and viable. The only phenotype observed under normal conditions was a slightly smaller proportion of B cells in splenocytes in DOCK10-/- mice than in the other mouse lines. We also found that the migration ability of macrophages is impaired in DOCK10-/- and DOCK11-/- mice and that the severity of experimental autoimmune encephalomyelitis was ameliorated only in DOCK10-/- mice. No apparent phenotype was observed for DOCK9-/- mice. Further investigations indicated that lipopolysaccharide stimulation up-regulates DOCK10 expression in microglia and that microglial migration is decreased in DOCK10-/- mice. Up-regulation of C-C motif chemokine ligand 2 (CCL2) expression induced by activation of Toll-like receptor 4 or 9 signaling was reduced in DOCK10-/- astrocytes compared with WT astrocytes. Taken together, our findings suggest that DOCK10 plays a role in innate immunity and neuroinflammation and might represent a potential therapeutic target for managing multiple sclerosis.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Esclerosis Múltiple/metabolismo , Animales , Astrocitos/patología , Modelos Animales de Enfermedad , Ratones , Microglía/patología , Esclerosis Múltiple/patología
5.
J Biol Chem ; 294(36): 13421-13433, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31337702

RESUMEN

Dedicator of cytokinesis 8 (DOCK8) is a guanine nucleotide exchange factor whose loss of function results in immunodeficiency, but its role in the central nervous system (CNS) has been unclear. Microglia are the resident immune cells of the CNS and are implicated in the pathogenesis of various neurodegenerative diseases, including multiple sclerosis (MS) and glaucoma, which affects the visual system. However, the exact roles of microglia in these diseases remain unknown. Herein, we report that DOCK8 is expressed in microglia but not in neurons or astrocytes and that its expression is increased during neuroinflammation. To define the role of DOCK8 in microglial activity, we focused on the retina, a tissue devoid of infiltrating T cells. The retina is divided into distinct layers, and in a disease model of MS/optic neuritis, DOCK8-deficient mice exhibited a clear reduction in microglial migration through these layers. Moreover, neuroinflammation severity, indicated by clinical scores, visual function, and retinal ganglion cell (RGC) death, was reduced in the DOCK8-deficient mice. Furthermore, using a glaucoma disease model, we observed impaired microglial phagocytosis of RGCs in DOCK8-deficient mice. Our data demonstrate that DOCK8 is expressed in microglia and regulates microglial activity in disease states. These findings contribute to a better understanding of the molecular pathways involved in microglial activation and implicate a role of DOCK8 in several neurological diseases.


Asunto(s)
Modelos Animales de Enfermedad , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Microglía/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Células Cultivadas , Femenino , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Am J Pathol ; 187(12): 2876-2885, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28919108

RESUMEN

The major role of the renin-angiotensin system (RAS), including that of angiotensin II (Ang II), the principal effector molecule, in the cardiovascular system is well known. Increasing evidence suggests that the RAS also plays a role in the development of autoimmune diseases. Optic neuritis (ie, inflammation of the optic nerve, with retinal ganglion cell loss) is strongly associated with multiple sclerosis. We investigated the effects of candesartan, an Ang II receptor antagonist, on optic neuritis in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. The Ang II concentration was increased in the early phase of EAE. Oral administration of candesartan markedly attenuated demyelination of the optic nerve and spinal cord and reduced retinal ganglion cell loss and visual impairment in mice with EAE. In vitro analyses revealed that Ang II up-regulated the expression of Toll-like receptor (TLR)-4 in astrocytes via the NF-κB pathway. In addition, Ang II treatment enhanced lipopolysaccharide-induced production of monocyte chemoattractant protein 1 in astrocytes, and pretreatment with candesartan or SN50, an NF-κB inhibitor, suppressed the effects of Ang II. The novel pathway of RAS-NF-κB-TLR4 in glial cells identified in the present study may be a valid therapeutic target for neurodegeneration in neuroinflammatory diseases.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Degeneración Nerviosa/patología , Neuritis Óptica/patología , Sistema Renina-Angiotensina/efectos de los fármacos , Animales , Bencimidazoles/farmacología , Compuestos de Bifenilo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones , Ratones Endogámicos C57BL , Sistema Renina-Angiotensina/fisiología , Tetrazoles/farmacología
7.
Am J Pathol ; 185(12): 3238-47, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26476348

RESUMEN

Brain-derived neurotrophic factor (BDNF) regulates neural cell survival mainly by activating TrkB receptors. Several lines of evidence support a key role for BDNF-TrkB signaling in survival of adult retinal ganglion cells in animal models of optic nerve injury (ONI), but the neuroprotective effect of exogenous BDNF is transient. Glial cells have recently attracted considerable attention as mediators of neural cell survival, and TrkB expression in retinal glia suggests its role in neuroprotection. To elucidate this point directly, we examined the effect of ONI on TrkB(flox/flox):glial fibrillary acidic protein (GFAP)-Cre+ (TrkB(GFAP)) knockout (KO) mice, in which TrkB is deleted in retinal glial cells. ONI markedly increased mRNA expression levels of basic fibroblast growth factor (bFGF) in wild-type (WT) mice but not in TrkB(GFAP) KO mice. Immunohistochemical analysis at 7 days after ONI (d7) revealed bFGF up-regulation mainly occurred in Müller glia. ONI-induced retinal ganglion cell loss in WT mice was consistently mild compared with TrkB(GFAP) KO mice at d7. On the other hand, ONI severely decreased TrkB expression in both WT and TrkB(GFAP) KO mice after d7, and the severity of retinal degeneration was comparable with TrkB(GFAP) KO mice at d14. Our data provide direct evidence that glial TrkB signaling plays an important role in the early stage of neural protection after traumatic injury.


Asunto(s)
Neuroglía/metabolismo , Neuroprotección/fisiología , Traumatismos del Nervio Óptico/metabolismo , Receptor trkB/fisiología , Animales , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factores de Crecimiento Nervioso/biosíntesis , Traumatismos del Nervio Óptico/complicaciones , Traumatismos del Nervio Óptico/patología , Degeneración Retiniana/etiología , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Transducción de Señal/fisiología
8.
Am J Pathol ; 185(3): 756-64, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25542970

RESUMEN

Valproic acid (VPA) is widely prescribed for treatment of epilepsy, mood disorders, migraines, and neuropathic pain. It exerts its therapeutic benefits through multiple mechanisms, including enhancement of GABAergic activity, activation of prosurvival protein kinases, and inhibition of histone deacetylase. Increasing evidence suggests that VPA possesses neuroprotective properties. We examined neuroprotective effects of VPA in an N-methyl-d-aspartate (NMDA) excitotoxicity model, which mimics some of the pathological features of glaucoma. In vivo retinal imaging using optical coherence tomography revealed that NMDA-induced retinal degeneration was suppressed in the VPA-treated retina, and histological analyses confirmed that VPA reduced retinal ganglion cell death. In vivo electrophysiological analyses demonstrated that visual impairment was prevented in the VPA-treated retina, clearly establishing both histological and functional effects of VPA. Brain-derived neurotrophic factor (BDNF) expression was up-regulated in Müller glial cells, and neuroprotective effects of VPA on retinal ganglion cells were significantly reduced in a conditional knockout mouse strain with deletion of tropomyosin receptor kinase B (TrkB), a receptor for BDNF from retinal ganglion cells. The results show that VPA stimulates BDNF up-regulation in Müller glial cells and provides direct evidence that neuronal TrkB is important in VPA-mediated neuroprotection. Also, VPA suppresses oxidative stress induced by NMDA in the retina. Our findings raise intriguing possibilities that the widely prescribed drug VPA may be useful for treatment of glaucoma.


Asunto(s)
Muerte Celular/efectos de los fármacos , N-Metilaspartato/farmacología , Fármacos Neuroprotectores/farmacología , Receptor trkB/metabolismo , Células Ganglionares de la Retina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Ratones , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Regulación hacia Arriba/efectos de los fármacos
9.
Int J Mol Sci ; 17(9)2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27657046

RESUMEN

Neurotrophic factors play key roles in the development and survival of neurons. The potent neuroprotective effects of neurotrophic factors, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), glial cell-line derived neurotrophic factor (GDNF) and nerve growth factor (NGF), suggest that they are good therapeutic candidates for neurodegenerative diseases. Glaucoma is a neurodegenerative disease of the eye that causes irreversible blindness. It is characterized by damage to the optic nerve, usually due to high intraocular pressure (IOP), and progressive degeneration of retinal neurons called retinal ganglion cells (RGCs). Current therapy for glaucoma focuses on reduction of IOP, but neuroprotection may also be beneficial. BDNF is a powerful neuroprotective agent especially for RGCs. Exogenous application of BDNF to the retina and increased BDNF expression in retinal neurons using viral vector systems are both effective in protecting RGCs from damage. Furthermore, induction of BDNF expression by agents such as valproic acid has also been beneficial in promoting RGC survival. In this review, we discuss the therapeutic potential of neurotrophic factors in retinal diseases and focus on the differential roles of glial and neuronal TrkB in neuroprotection. We also discuss the role of neurotrophic factors in neuroregeneration.

10.
Biochem Biophys Rep ; 39: 101799, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39161576

RESUMEN

Neurodegenerative diseases including glaucoma affect insulin signaling, and insulin treatment has been shown to reverse the neurodegenerative loss of dendritic complexity in retinal ganglion cells. Therefore, strategies for enhancing or maintaining insulin signaling are worth pursuing to establish new therapies for these diseases. In the present study, we generated constitutively active insulin receptor (F-iIR) and insulin-like growth factor-1 receptor (F-iIGF1R) using a system that forces membrane localization of the intracellular domains of these receptors by farnesylation. Immunohistochemistry and Western blot analysis revealed that F-iIR and F-iIGF1R caused the activation of ERK and AKT in the absence of ligands in vitro. Our results suggest that in vivo effects of F-iIR and F-iIGF1R on the progression of neurodegenerative diseases should be investigated in the future.

11.
J Neurosci ; 32(1): 264-74, 2012 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-22219288

RESUMEN

Dock3, a new member of the guanine nucleotide exchange factors, causes cellular morphological changes by activating the small GTPase Rac1. Overexpression of Dock3 in neural cells promotes axonal outgrowth downstream of brain-derived neurotrophic factor (BDNF) signaling. We previously showed that Dock3 forms a complex with Fyn and WASP (Wiskott-Aldrich syndrome protein) family verprolin-homologous (WAVE) proteins at the plasma membrane, and subsequent Rac1 activation promotes actin polymerization. Here we show that Dock3 binds to and inactivates glycogen synthase kinase-3ß (GSK-3ß) at the plasma membrane, thereby increasing the nonphosphorylated active form of collapsin response mediator protein-2 (CRMP-2), which promotes axon branching and microtubule assembly. Exogenously applied BDNF induced the phosphorylation of GSK-3ß and dephosphorylation of CRMP-2 in hippocampal neurons. Moreover, increased phosphorylation of GSK-3ß was detected in the regenerating axons of transgenic mice overexpressing Dock3 after optic nerve injury. These results suggest that Dock3 plays important roles downstream of BDNF signaling in the CNS, where it regulates cell polarity and promotes axonal outgrowth by stimulating dual pathways: actin polymerization and microtubule assembly.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Proteínas Portadoras/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Conos de Crecimiento/metabolismo , Hipocampo/metabolismo , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células COS , Chlorocebus aethiops , Glucógeno Sintasa Quinasa 3 beta , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/ultraestructura , Factores de Intercambio de Guanina Nucleótido , Células HEK293 , Hipocampo/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura
12.
Genes Cells ; 17(8): 688-97, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22734669

RESUMEN

Dock3, a new member of the guanine nucleotide exchange factor family, causes cellular morphological changes by activating the small GTPase Rac1. Overexpression of Dock3 in neural cells promotes neurite outgrowth through the formation of a protein complex with Fyn and WAVE downstream of brain-derived neurotrophic factor (BDNF) signaling. Here, we report a novel Dock3-mediated BDNF pathway for neurite outgrowth. We show that Dock3 forms a complex with Elmo and activated RhoG downstream of BDNF-TrkB signaling and induces neurite outgrowth via Rac1 activation in PC12 cells. We also show the importance of Dock3 phosphorylation in Rac1 activation and show two key events that are necessary for efficient Dock3 phosphorylation: membrane recruitment of Dock3 and interaction of Dock3 with Elmo. These results suggest that Dock3 plays important roles downstream of BDNF signaling in the central nervous system where it stimulates actin polymerization by multiple pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Receptor trkB/metabolismo , Transducción de Señal , Factores Complejos Ternarios/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Células COS , Proteínas Portadoras/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Chlorocebus aethiops , Activación Enzimática , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Factores de Intercambio de Guanina Nucleótido , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Neuritas/efectos de los fármacos , Neuritas/fisiología , Neuropéptidos/genética , Neuropéptidos/metabolismo , Células PC12 , Fosforilación , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Ratas , Receptor trkB/genética , Factores Complejos Ternarios/genética , Transfección , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1 , Proteínas de Unión al GTP rho
13.
Proc Natl Acad Sci U S A ; 107(16): 7586-91, 2010 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-20368433

RESUMEN

Atypical Rho-guanine nucleotide exchange factors (Rho-GEFs) that contain Dock homology regions (DHR-1 and DHR-2) are expressed in a variety of tissues; however, their functions and mechanisms of action remain unclear. We identify key conserved amino acids in the DHR-2 domain that are critical for the catalytic activity of Dock-GEFs (Dock1-4). We further demonstrate that Dock-GEFs directly associate with WASP family verprolin-homologous (WAVE) proteins through the DHR-1 domain. Brain-derived neurotrophic factor (BDNF)-TrkB signaling recruits the Dock3/WAVE1 complex to the plasma membrane, whereupon Dock3 activates Rac and dissociates from the WAVE complex in a phosphorylation-dependent manner. BDNF induces axonal sprouting through Dock-dependent Rac activation, and adult transgenic mice overexpressing Dock3 exhibit enhanced optic nerve regeneration after injury without affecting WAVE expression levels. Our results highlight a unique mechanism through which Dock-GEFs achieve spatial and temporal restriction of WAVE signaling, and identify Dock-GEF activity as a potential therapeutic target for axonal regeneration.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas del Tejido Nervioso/fisiología , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células COS , Chlorocebus aethiops , Hipocampo/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Regeneración Nerviosa , Neuronas/metabolismo , Nervio Óptico/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho , Homología de Secuencia de Aminoácido , Transducción de Señal
14.
Neurosci Lett ; 799: 137124, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36780941

RESUMEN

Visual disturbance after optic nerve injury is a serious problem. Attempts have been made to enhance the intrinsic ability of retinal ganglion cells (RGCs) to regenerate their axons, and the importance of PI3K/Akt and RAF/MEK/ERK signal activation has been suggested. Since these signals are shared with oncogenic signaling cascades, in this study, we focused on a constitutively active form of K-Ras, K-RasV12, to determine if overexpression of this molecule could stimulate axon regeneration. We confirmed that K-RasV12 phosphorylated Akt and ERK in vitro. Intravitreal delivery of AAV2-K-RasV12 increased the number of surviving RGCs and promoted 1.0 mm of axon regeneration one week after optic nerve injury without inducing abnormal proliferative effects in the RGCs. In addition, AAV2-K-RasV12 induced robust RGC axon regeneration, reaching as far as approximately 2.5 mm from the injury site, in eight weeks. Our findings suggest that AAV2-K-RasV12 could provide a good model for speedy and efficient analysis of the mechanism underlying axon regeneration in vivo.


Asunto(s)
Traumatismos del Nervio Óptico , Humanos , Axones/fisiología , Proteínas Proto-Oncogénicas c-akt , Fosfatidilinositol 3-Quinasas , Regeneración Nerviosa/fisiología
15.
Cell Death Discov ; 9(1): 166, 2023 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-37188749

RESUMEN

Dedicator of cytokinesis 3 (DOCK3) is an atypical member of the guanine nucleotide exchange factors (GEFs) and plays important roles in neurite outgrowth. DOCK3 forms a complex with Engulfment and cell motility protein 1 (Elmo1) and effectively activates Rac1 and actin dynamics. In this study, we screened 462,169 low-molecular-weight compounds and identified the hit compounds that stimulate the interaction between DOCK3 and Elmo1, and neurite outgrowth in vitro. Some of the derivatives from the hit compound stimulated neuroprotection and axon regeneration in a mouse model of optic nerve injury. Our findings suggest that the low-molecular-weight DOCK3 activators could be a potential therapeutic candidate for treating axonal injury and neurodegenerative diseases including glaucoma.

16.
Biochem Biophys Rep ; 29: 101197, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35028438

RESUMEN

Lighting conditions may affect the development of retinal degenerative diseases such as macular degeneration. In this study, to determine whether the lighting environment affects the progression of degeneration of retinal ganglion cells (RGCs), we examined glutamate/aspartate transporter (GLAST) heterozygous (GLAST+/-) mice, a mouse model of normal tension glaucoma. GLAST+/- mice were reared under a 12-h light-dark cycle (Light/Dark) or complete darkness (Dark/Dark) condition after birth. The total RGC number in the Dark/Dark group was significantly decreased compared with the Light/Dark group at 3 weeks old, while the number of osteopontin-positive αRGCs were similar in both groups. At 6 and 12 weeks old, the total RGC number were not significantly different in both conditions. In addition, the retinal function examined by multifocal electroretinogram were similar at 12 weeks old. These results suggest that lighting conditions may regulate the progression of RGC degeneration in some types of glaucoma.

17.
J Clin Invest ; 117(7): 1763-70, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17607354

RESUMEN

Glaucoma, a progressive optic neuropathy due to retinal ganglion cell (RGC) degeneration, is one of the leading causes of irreversible blindness. Although glaucoma is often associated with elevated intraocular pressure (IOP), IOP elevation is not detected in a significant subset of glaucomas, such as normal tension glaucoma (NTG). Moreover, in some glaucoma patients, significant IOP reduction does not prevent progression of the disease. Thus, understanding IOP-independent mechanisms of RGC loss is important. Here, we show that mice deficient in the glutamate transporters GLAST or EAAC1 demonstrate spontaneous RGC and optic nerve degeneration without elevated IOP. In GLAST-deficient mice, the glutathione level in Müller glia was decreased; administration of glutamate receptor blocker prevented RGC loss. In EAAC1-deficient mice, RGCs were more vulnerable to oxidative stress. These findings suggest that glutamate transporters are necessary both to prevent excitotoxic retinal damage and to synthesize glutathione, a major cellular antioxidant and tripeptide of glutamate, cysteine, and glycine. We believe these mice are the first animal models of NTG that offer a powerful system for investigating mechanisms of neurodegeneration in NTG and developing therapies directed at IOP-independent mechanisms of RGC loss.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/metabolismo , Modelos Animales de Enfermedad , Glaucoma/metabolismo , Glaucoma/patología , Presión Intraocular , Sistema de Transporte de Aminoácidos X-AG/deficiencia , Sistema de Transporte de Aminoácidos X-AG/genética , Animales , Regulación de la Expresión Génica , Glaucoma/genética , Ácido Glutámico/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Enfermedades del Nervio Óptico/genética , Enfermedades del Nervio Óptico/metabolismo , Enfermedades del Nervio Óptico/patología , Estrés Oxidativo , Células Ganglionares de la Retina/metabolismo , Visión Ocular
18.
Antioxidants (Basel) ; 9(9)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32947996

RESUMEN

Glaucoma is a neurodegenerative disease of the eye, which involves degeneration of retinal ganglion cells (RGCs): the output neurons of the retina to the brain, which with their axons comprise the optic nerve. Recent studies have shown the possible involvement of oxidative stress in the pathogenesis of glaucoma, especially in the subtype of normal tension glaucoma. Basic experiments utilizing rodent and primate models of glaucoma revealed that antioxidants protect RGCs under various pathological conditions including glutamate neurotoxicity and optic nerve injury. These results suggested that existing drugs and food factors may be useful for prevention and hence therapy of glaucoma. In this review, we highlight some therapeutic candidates, particularly those with antioxidant properties, and discuss the therapeutic potential of RGC protection by modulating gene expressions that prevent and ameliorate glaucoma.

19.
Sci Rep ; 10(1): 15709, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32973242

RESUMEN

Optic nerve injury induces optic nerve degeneration and retinal ganglion cell (RGC) death that lead to visual disturbance. In this study, we examined if topical ripasudil has therapeutic potential in adult mice after optic nerve crush (ONC). Topical ripasudil suppressed ONC-induced phosphorylation of p38 mitogen-activated protein kinase and ameliorated RGC death. In addition, topical ripasudil significantly suppressed the phosphorylation of collapsin response mediator protein 2 and cofilin, and promoted optic nerve regeneration. These results suggest that topical ripasudil promotes RGC protection and optic nerve regeneration by modulating multiple signaling pathways associated with neural cell death, microtubule assembly and actin polymerization.


Asunto(s)
Axones/efectos de los fármacos , Isoquinolinas/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Traumatismos del Nervio Óptico/tratamiento farmacológico , Sulfonamidas/uso terapéutico , Animales , Axones/fisiología , Isoquinolinas/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Compresión Nerviosa , Regeneración Nerviosa/fisiología , Fármacos Neuroprotectores/administración & dosificación , Traumatismos del Nervio Óptico/metabolismo , Traumatismos del Nervio Óptico/fisiopatología , Fosforilación/efectos de los fármacos , Células Ganglionares de la Retina/efectos de los fármacos , Células Ganglionares de la Retina/metabolismo , Sulfonamidas/administración & dosificación
20.
Br J Ophthalmol ; 103(2): 161-166, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30366949

RESUMEN

Glaucoma is one of the leading causes of vision loss in the world. Currently, pharmacological intervention for glaucoma therapy is limited to eye drops that reduce intraocular pressure (IOP). Recent studies have shown that various factors as well as IOP are involved in the pathogenesis of glaucoma, especially in the subtype of normal tension glaucoma. To date, various animal models of glaucoma have been established, including glutamate/aspartate transporter knockout (KO) mice, excitatory amino acid carrier 1 KO mice, optineurin E50K knock-in mice, DBA/2J mice and experimentally induced models. These animal models are very useful for elucidating the pathogenesis of glaucoma and for identifying potential therapeutic targets. However, each model represents only some aspects of glaucoma, never the whole disease. This review will summarise the benefits and limitations of using disease models of glaucoma and recent basic research in retinal protection using existing drugs.


Asunto(s)
Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Glaucoma/tratamiento farmacológico , Animales , Antihipertensivos/uso terapéutico , Humanos , Preparaciones Farmacéuticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA